1
|
Xu J, Shepard BD, Pluznick JL. Roles of sensory receptors in non-sensory organs: the kidney and beyond. Nat Rev Nephrol 2025; 21:253-263. [PMID: 39753689 PMCID: PMC11929601 DOI: 10.1038/s41581-024-00917-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 02/02/2025]
Abstract
Olfactory receptors (ORs), taste receptors and opsins are well-known for their pivotal roles in mediating the senses of smell, taste and sight, respectively. However, in the past two decades, research has shown that these sensory receptors also regulate physiological processes in a variety of non-sensory tissues. Although ORs, taste receptors and opsins have all been shown to have physiological roles beyond their traditional locations, most work in the kidney has focused on ORs. To date, renal ORs have been shown to have roles in blood pressure regulation (OLFR78 and OLFR558) and glucose homeostasis (OLFR1393). However, sensory receptors remain drastically understudied outside of traditional sensory systems, in part because of inherent challenges in studying these receptors. Increased knowledge of the physiological and pathophysiological roles of sensory receptors has the potential to substantially improve understanding of the function of numerous organs and systems, including the kidney. In addition, most sensory receptors are G protein-coupled receptors, which are considered to be the most druggable class of proteins, and thus could potentially be exploited as future therapeutic targets.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Blythe D Shepard
- Department of Human Science, Georgetown University, Washington, DC, USA
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Kachi E, Kazama K, Murakami M, Onda K. Opsin 3, encoding a non-visual photoreceptor, is a pseudogene in cattle. Res Vet Sci 2025; 186:105586. [PMID: 39985965 DOI: 10.1016/j.rvsc.2025.105586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
The non-visual photoreceptor opsin 3 (OPN3) performs various functions by directly accepting light and activating G protein-coupled receptor signalling in non-visual tissues. OPN3, which is expressed in brown adipocytes, induces the expression of uncoupling protein 1 to generate heat upon exposure to blue light. There have been few reports on OPN3 expression in ruminants, and the mRNA sequence registered in a database as bovine OPN3 is the only predicted sequence. Therefore, we examined OPN3 expression in cattle. In the bovine retina, amplified fragments of the visual opsins, rhodopsin and opsin 1 and the non-visual opsins, opsin 4 and opsin 5, were detected using reverse transcription PCR, whereas no amplified PCR products of OPN3 were detected. Eleven tissues were used to analyse OPN3 expression in the entire body of bovines, including the testis and placenta, where OPN3 is highly expressed in humans and mice. No amplified PCR products were detected in any of the tissues. In a comparison among animal species, OPN3 was found to be expressed in mouse and horse testes, whereas no expression was observed in cattle and goat tissues. RNA sequencing revealed no OPN3 transcripts in the bovine placenta. Direct genomic DNA sequencing revealed a stop codon in the middle of the bovine OPN3 sequence. These results suggest that bovine OPN3 is pseudogenic as it does not encode a complete receptor protein. They also indicated that OPN3 is pseudogenic not only in cattle but also in other Cetartiodactyla, including goats.
Collapse
Affiliation(s)
- Erina Kachi
- Azabu University, Graduate School of Veterinary Medicine, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan.
| | - Kei Kazama
- Azabu University, Graduate School of Veterinary Medicine, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan.
| | - Masaru Murakami
- Azabu University, Graduate School of Veterinary Medicine, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan.
| | - Ken Onda
- Azabu University, Graduate School of Veterinary Medicine, 1-17-71, Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201, Japan.
| |
Collapse
|
3
|
Liu Y, Zhao Y, Zhang J, Wu X, Li Y, Tang K, Han Z. OPN3 enhances the proliferation, migration, and invasion of triple-negative breast cancer cells via the regulation of the TGF-β signaling pathway. Transl Cancer Res 2025; 14:1141-1156. [PMID: 40104724 PMCID: PMC11912049 DOI: 10.21037/tcr-24-1374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/17/2024] [Indexed: 03/20/2025]
Abstract
Background Opsin3 (OPN3) belongs to the guanine nucleotide-binding protein-coupled receptor superfamily, implicated in several pathological mechanisms that contribute to tumor development and resistance to treatment. This study examined OPN3 expression in triple-negative breast cancer (TNBC) tissues and its function in TNBC cell propagation, invasion, and migration. Methods The study analyzed OPN3 expression in TNBC patients and its diagnostic value using immunohistochemistry (IHC) staining and The Cancer Genome Atlas (TCGA) data. To evaluate the impact of OPN3 on the growth, invasion, and migration of TNBC cells, both in vitro and in vivo experiments were carried out. The biological mechanisms of OPN3-induced cell migration and invasion were evaluated via quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting (WB) analyses. Results In this study, OPN3 upregulation in TNBC tissues was found to be correlated with decreased overall survival (OS) and progression-free survival (PFS) rates. The propagation, invasion, and migration of BT-549 cells were promoted and apoptosis was reduced after OPN3 overexpression. It was silenced in BT-549 cells, which resulted in the opposite effects. Further, the transforming growth factor-beta (TGF-β)/SMAD2 signaling pathway was stimulated and the epithelial-mesenchymal transition (EMT) was modulated by OPN3 overexpression in BT-549 cells. Conclusions The findings revealed that OPN3 is involved in the stimulation of the TGF-β/SMAD2 signaling pathway, which promotes the growth, migration, and dissemination of TNBC cells. Moreover, OPN3 can serve as a diagnostic biomarker and a treatment target for TNBC.
Collapse
Affiliation(s)
- Yameng Liu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Yue Zhao
- Departments of Oncology Gynecology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jingjing Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Xiao Wu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Yumei Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Ke Tang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, China
| | - Zhengquan Han
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
4
|
Haddad HK, Mercado-Reyes JI, Mustafá ER, D’Souza SP, Chung CS, Nestor RRM, Olinski LE, Martinez Damonte V, Saskin J, Vemaraju S, Raingo J, Kauer JA, Lang RA, Oancea E. Hypothalamic opsin 3 suppresses MC4R signaling and potentiates Kir7.1 to promote food consumption. Proc Natl Acad Sci U S A 2025; 122:e2403891122. [PMID: 39951488 PMCID: PMC11874419 DOI: 10.1073/pnas.2403891122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 12/02/2024] [Indexed: 02/16/2025] Open
Abstract
Mammalian opsin 3 (OPN3) is a member of the opsin family of G-protein-coupled receptors with ambiguous light sensitivity. OPN3 was first identified in the brain (and named encephalopsin) and subsequently found to be expressed in other tissues. In adipocytes, OPN3 is necessary for light responses that modulate lipolysis and glucose uptake, while OPN3 in human skin melanocytes regulates pigmentation in a light-independent manner. Despite its initial discovery in the brain, OPN3 functional mechanisms in the brain remain elusive. Here, we investigated the molecular mechanism of OPN3 function in the paraventricular nucleus (PVN) of the hypothalamus. We show that Opn3 is coexpressed with the melanocortin 4 receptor (Mc4r) in a population of PVN neurons, where it negatively regulates MC4R-mediated cAMP signaling in a specific and Gαi/o-dependent manner. Under baseline conditions, OPN3 via Gαi/o potentiates the activity of the inward rectifying Kir7.1 channel, previously shown to be closed in response to agonist-mediated activation of MC4R in a Gαs-independent manner. In mice, we found that Opn3 in Mc4r-expressing neurons regulates food consumption. Our results reveal the first mechanistic insight into OPN3 function in the hypothalamus, uncovering a unique mechanism by which OPN3 functions to potentiate Kir7.1 activity and negatively regulate MC4R-mediated cAMP signaling, thereby promoting food intake.
Collapse
Affiliation(s)
- Hala K. Haddad
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Jonathan I. Mercado-Reyes
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - E. Román Mustafá
- Electrophysiology Lab, Instituto Multidisciplinario de Biología Celular, La Plata, Buenos Aires1900, Argentina
| | - Shane P. D’Souza
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - C. Sean Chung
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Ramses R. M. Nestor
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Lauren E. Olinski
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Valentina Martinez Damonte
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Joshua Saskin
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| | - Shruti Vemaraju
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
| | - Jesica Raingo
- Electrophysiology Lab, Instituto Multidisciplinario de Biología Celular, La Plata, Buenos Aires1900, Argentina
| | - Julie A. Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA94305
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Abrahamson Pediatric Eye Institute, and Science of Light Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229
- Department of Ophthalmology, University of Cincinnati, Cincinnati, OH45229
| | - Elena Oancea
- Division of Biology and Medicine, Department of Neuroscience, Brown University, Providence, RI02912
| |
Collapse
|
5
|
Eltanahy AM, Aupetit A, Buhr ED, Van Gelder RN, Gonzales AL. Light-sensitive Ca 2+ signaling in the mammalian choroid. Proc Natl Acad Sci U S A 2024; 121:e2418429121. [PMID: 39514305 PMCID: PMC11573543 DOI: 10.1073/pnas.2418429121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
The choroid is the thin, vasculature-filled layer of the eye situated between the sclera and the retina, where it serves the metabolic needs of the light-sensing photoreceptors in the retina. Illumination of the interior surface of the back of the eye (fundus) is a critical regulator of subretinal fluid homeostasis, which determines the overall shape of the eye, but it is also important for choroidal perfusion. Noted for having some of the highest blood flow rates in the body, the choroidal vasculature has been reported to lack intrinsic, intravascular pressure-induced (myogenic) autoregulatory mechanisms. Here, we ask how light directly regulates choroid perfusion and ocular fluid homeostasis, testing the hypothesis that light facilitates ocular fluid absorption by directly increasing choroid endothelial permeability and decreasing choroid perfusion. Utilizing ex vivo pressurized whole-choroid and whole-eye preparations from mice expressing cell-specific Ca2+ indicators, we found that the choroidal vasculature has two intrinsically light-sensitive Ca2+-signaling mechanisms: One increases Ca2+-dependent production of nitric oxide in choroidal endothelial cells; the other promotes vasoconstriction through Ca2+ elevation in vascular smooth muscle cells. In addition, we found that choroidal flow, or pressure, modulates endothelial and smooth muscle photosensitivity and trans-retinal absorption of fluid into the choroid. These results collectively suggest that the choroid vasculature exhibits an inverted form of autoregulatory control, where pressure- and light-induced mechanisms work in opposition to regulate blood flow and maintain fluid balance in response to changes in light and dark, aligning with the metabolic needs of photoreceptors.
Collapse
Affiliation(s)
- Ahmed M Eltanahy
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318
| | - Alex Aupetit
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318
| | - Ethan D Buhr
- Department of Ophthalmology, University of Washington, Seattle, WA 98104
- Roger and Angie Karalis Retina Center, Department of Ophthalmology, University of Washington, Seattle, WA 98104
| | - Russell N Van Gelder
- Department of Ophthalmology, University of Washington, Seattle, WA 98104
- Roger and Angie Karalis Retina Center, Department of Ophthalmology, University of Washington, Seattle, WA 98104
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
- Department of Neurobiology & Biophysics, University of Washington, Seattle, WA 98104
| | - Albert L Gonzales
- Department of Physiology and Cell Biology, Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno School of Medicine, Reno, NV 89557-0318
| |
Collapse
|
6
|
Zhao C, Bo J, Li T, Tian J, Long T, He Y, Chen S, Liu C. Blue light-driven cell cycle arrest in thyroid cancer via Retinal-OPN3 complex. Cell Commun Signal 2024; 22:530. [PMID: 39487504 PMCID: PMC11531186 DOI: 10.1186/s12964-024-01908-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is the most common type of thyroid malignancy, with a rising incidence. Traditional treatments, such as thyroidectomy and radiotherapy, often lead to significant side effects, including impaired thyroid function. Therefore, there is an urgent need for non-invasive therapeutic approaches. This study aims to explore the potential of photobiomodulation therapy (PBMT), a non-invasive treatment using specific wavelengths of light, in the management of PTC. METHODS We investigated the effects of blue light PBMT on PTC cells, focusing on the Retinal-OPSIN 3 (OPN3) complex's role in mediating cellular responses. Blue light exposure was applied to PTC cells, and subsequent changes in cellular proliferation, cell cycle progression, and protein expression were analyzed. Statistical tests, including one-way ANOVA and t-tests, were used to evaluate the significance of the findings. RESULTS Blue light exposure led to the dissociation of 11-cis-retinal from OPN3, resulting in the accumulation of all-trans retinal. This accumulation disrupted cellular proliferation pathways and induced G0/G1 cell cycle arrest in PTC cells. The Retinal-OPN3 complex was found to be a key mediator in these processes, demonstrating that thyroid cells can respond to specific light wavelengths and utilize their photoreceptive potential for therapeutic purposes. CONCLUSIONS Our findings suggest that PBMT, through the modulation of the Retinal-OPN3 complex, offers a promising non-invasive approach for treating PTC. This study highlights the therapeutic potential of light signal transduction in non-ocular tissues and opens new avenues for non-invasive cancer therapies.
Collapse
Affiliation(s)
- Changrui Zhao
- Department of Endocrinology, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiaqiang Bo
- Department of Endocrinology, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Tianyu Li
- Department of Endocrinology, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiameng Tian
- Department of Endocrinology, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Tian Long
- Mudi Meng Honors College, China Pharmaceutical University, Nanjing, 211198, China
| | - Yingying He
- Department of Endocrinology, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Siyu Chen
- Department of Endocrinology, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Chang Liu
- Department of Endocrinology, Nanjing Drum Tower Hospital, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China.
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases (China Pharmaceutical University), Nanjing, China.
| |
Collapse
|
7
|
Wang CH, Tsuji T, Wu LH, Yang CY, Huang TL, Sato M, Shamsi F, Tseng YH. Endothelin 3/EDNRB signaling induces thermogenic differentiation of white adipose tissue. Nat Commun 2024; 15:7215. [PMID: 39174539 PMCID: PMC11341701 DOI: 10.1038/s41467-024-51579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
Thermogenic adipose tissue, consisting of brown and beige fat, regulates nutrient utilization and energy metabolism. Human brown fat is relatively scarce and decreases with obesity and aging. Hence, inducing thermogenic differentiation of white fat offers an attractive way to enhance whole-body metabolic capacity. Here, we show the role of endothelin 3 (EDN3) and endothelin receptor type B (EDNRB) in promoting the browning of white adipose tissue (WAT). EDNRB overexpression stimulates thermogenic differentiation of human white preadipocytes through cAMP-EPAC1-ERK activation. In mice, cold induces the expression of EDN3 and EDNRB in WAT. Deletion of EDNRB in adipose progenitor cells impairs cold-induced beige adipocyte formation in WAT, leading to excessive weight gain, glucose intolerance, and insulin resistance upon high-fat feeding. Injection of EDN3 into WAT promotes browning and improved whole-body glucose metabolism. The findings shed light on the mechanism of WAT browning and offer potential therapeutics for obesity and metabolic disorders.
Collapse
Affiliation(s)
- Chih-Hao Wang
- Graduate Institute of Cell Biology, China Medical University, Taichung City, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taiwan.
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Tadataka Tsuji
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Li-Hong Wu
- Graduate Institute of Cell Biology, China Medical University, Taichung City, Taiwan
| | - Cheng-Ying Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taiwan
| | - Tian Lian Huang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Mari Sato
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
8
|
Tsuji T, Tolstikov V, Zhang Y, Huang TL, Camara H, Halpin M, Narain NR, Yau KW, Lynes MD, Kiebish MA, Tseng YH. Light-responsive adipose-hypothalamus axis controls metabolic regulation. Nat Commun 2024; 15:6768. [PMID: 39117652 PMCID: PMC11310318 DOI: 10.1038/s41467-024-50866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Light is fundamental for biological life, with most mammals possessing light-sensing photoreceptors in various organs. Opsin3 is highly expressed in adipose tissue which has extensive communication with other organs, particularly with the brain through the sympathetic nervous system (SNS). Our study reveals a new light-triggered crosstalk between adipose tissue and the hypothalamus. Direct blue-light exposure to subcutaneous white fat improves high-fat diet-induced metabolic abnormalities in an Opsin3-dependent manner. Metabolomic analysis shows that blue light increases circulating levels of histidine, which activates histaminergic neurons in the hypothalamus and stimulates brown adipose tissue (BAT) via SNS. Blocking central actions of histidine and denervating peripheral BAT blunts the effects of blue light. Human white adipocytes respond to direct blue light stimulation in a cell-autonomous manner, highlighting the translational relevance of this pathway. Together, these data demonstrate a light-responsive metabolic circuit involving adipose-hypothalamus communication, offering a potential strategy to alleviate obesity-induced metabolic abnormalities.
Collapse
Affiliation(s)
- Tadataka Tsuji
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | - Yang Zhang
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Tian Lian Huang
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Henrique Camara
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Meghan Halpin
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | - King-Wai Yau
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew D Lynes
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | | | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
9
|
Cui Y, Auclair H, He R, Zhang Q. GPCR-mediated regulation of beige adipocyte formation: Implications for obesity and metabolic health. Gene 2024; 915:148421. [PMID: 38561165 DOI: 10.1016/j.gene.2024.148421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Obesity and its associated complications pose a significant burden on health. The non-shivering thermogenesis (NST) and metabolic capacity properties of brown adipose tissue (BAT), which are distinct from those of white adipose tissue (WAT), in combating obesity and its related metabolic diseases has been well documented. However, beige adipose tissue, the third and relatively novel type of adipose tissue, which emerges in extensive presence of WAT and shares similar favorable metabolic properties with BAT, has garnered considerable attention in recent years. In this review, we focused on the role of G protein-coupled receptors (GPCRs), the largest receptor family and the most successful class of drug targets in humans, in the induction of beige adipocytes. More importantly, we highlight researchers' clinical treatment attempts to ameliorate obesity and other related metabolic diseases through the formation and activation of beige adipose tissue. In summary, this review provides valuable insights into the formation of beige adipose tissue and the involvement of GPCRs, based on the latest advancements in scientific research.
Collapse
Affiliation(s)
- Yuanxu Cui
- Animal Zoology Department, Kunming Medical University, Kunming, China; Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, China
| | - Hugo Auclair
- Faculty of Medicine, François-Rabelais University, Tours, France
| | - Rong He
- Animal Zoology Department, Kunming Medical University, Kunming, China
| | - Qiang Zhang
- Animal Zoology Department, Kunming Medical University, Kunming, China.
| |
Collapse
|
10
|
Rao F, Xue T. Circadian-independent light regulation of mammalian metabolism. Nat Metab 2024; 6:1000-1007. [PMID: 38831000 DOI: 10.1038/s42255-024-01051-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/16/2024] [Indexed: 06/05/2024]
Abstract
The daily light-dark cycle is a key zeitgeber (time cue) for entraining an organism's biological clock, whereby light sensing by retinal photoreceptors, particularly intrinsically photosensitive retinal ganglion cells, stimulates the suprachiasmatic nucleus of the hypothalamus, a central pacemaker that in turn orchestrates the rhythm of peripheral metabolic activities. Non-rhythmic effects of light on metabolism have also been long known, and their transduction mechanisms are only beginning to unfold. Here, we summarize emerging evidence that, in mammals, light exposure or deprivation profoundly affects glucose homeostasis, thermogenesis and other metabolic activities in a clock-independent manner. Such light regulation could involve melanopsin-based, intrinsically photosensitive retinal ganglion cell-initiated brain circuits via the suprachiasmatic nucleus of the hypothalamus and other nuclei, or direct stimulation of opsins expressed in the hypothalamus, adipose tissue, blood vessels and skin to regulate sympathetic tone, lipolysis, glucose uptake, mitochondrial activation, thermogenesis, food intake, blood pressure and melanogenesis. These photic signalling events may coordinate with circadian-based mechanisms to maintain metabolic homeostasis, with dysregulation of this system underlying metabolic diseases caused by aberrant light exposure, such as environmental night light and shift work.
Collapse
Affiliation(s)
- Feng Rao
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Tian Xue
- Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
11
|
Deng X, Peng D, Yao Y, Huang K, Wang J, Ma Z, Fu J, Xu Y. Optogenetic therapeutic strategies for diabetes mellitus. J Diabetes 2024; 16:e13557. [PMID: 38751366 PMCID: PMC11096815 DOI: 10.1111/1753-0407.13557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 05/18/2024] Open
Abstract
Diabetes mellitus (DM) is a common chronic disease affecting humans globally. It is characterized by abnormally elevated blood glucose levels due to the failure of insulin production or reduction of insulin sensitivity and functionality. Insulin and glucagon-like peptide (GLP)-1 replenishment or improvement of insulin resistance are the two major strategies to treat diabetes. Recently, optogenetics that uses genetically encoded light-sensitive proteins to precisely control cell functions has been regarded as a novel therapeutic strategy for diabetes. Here, we summarize the latest development of optogenetics and its integration with synthetic biology approaches to produce light-responsive cells for insulin/GLP-1 production, amelioration of insulin resistance and neuromodulation of insulin secretion. In addition, we introduce the development of cell encapsulation and delivery methods and smart bioelectronic devices for the in vivo application of optogenetics-based cell therapy in diabetes. The remaining challenges for optogenetics-based cell therapy in the clinical translational study are also discussed.
Collapse
Affiliation(s)
- Xin Deng
- Department of EndocrinologyChildren's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio‐Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine for Clinical Evaluation and Translational ResearchZhejiang UniversityHangzhouChina
| | - Dandan Peng
- Department of EndocrinologyChildren's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
| | - Yuanfa Yao
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio‐Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine for Clinical Evaluation and Translational ResearchZhejiang UniversityHangzhouChina
| | - Ke Huang
- Department of EndocrinologyChildren's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
| | - Jinling Wang
- Department of EndocrinologyChildren's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
| | - Zhihao Ma
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio‐Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine for Clinical Evaluation and Translational ResearchZhejiang UniversityHangzhouChina
| | - Junfen Fu
- Department of EndocrinologyChildren's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
| | - Yingke Xu
- Department of EndocrinologyChildren's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio‐Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine for Clinical Evaluation and Translational ResearchZhejiang UniversityHangzhouChina
- Binjiang Institute of Zhejiang UniversityHangzhouChina
| |
Collapse
|
12
|
Zhang Z, Shi C, Han J, Ge X, Li N, Liu Y, Huang J, Chen S. Nonvisual system-mediated body color change in fish reveals nonvisual function of Opsin 3 in skin. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 252:112861. [PMID: 38335869 DOI: 10.1016/j.jphotobiol.2024.112861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
Body-color changes in many poikilothermic animals can occur quickly. This color change is generally initiated by visual system, followed by neuromuscular or neuroendocrine control. We have previously showed that the ventral skin color of the large yellow croaker (Larimichthys crocea) presents golden yellow in dark environment and quickly changes to silvery white in light environment. In the present study, we found that the light-induced whitening of ventral skin color was independent of visual input. Using light-emitting diode sources of different wavelength with same luminance (150 lx) but different absolute irradiance (0.039-0.333 mW/cm2), we further found that the blue light (λmax = 480 nm, 0.107 mW/cm2) is more effectively in induction of whitening of ventral skin color in compare with other light sources. Interestingly, the result of RT-PCR showed opsin 3 transcripts expressed in xanthophores. Recombinant protein of Opsin 3 with 11-cis retinal formed functional blue-sensitive pigment, with an absorption maximum at 468 nm. The HEK293T cells transfected with Opsin 3 showed a blue light-evoked Ca2+ response. Knock-down of Opsin 3 expression blocked the light-induced xanthosomes aggregation in vitro. Moreover, the light-induced xanthosomes aggregation was mediated via Ca2+-PKC and Ca2+-CaMKII pathways, and relied on microtubules and dynein. Decrease of cAMP levels was a prerequisite for xanthosomes aggregation. Our results provide a unique organism model exhibiting light-induced quick body color change, which was independent of visual input but rather rely on non-visual function of Opsin 3 within xanthophore.
Collapse
Affiliation(s)
- Zihao Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Chenchen Shi
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jian Han
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China; Key laboratory of fish applied biology and aquaculture in North China, College of Fisheries and Life Science, Dalian Ocean University, Dalian, Liaoning 116023, China
| | - Xiaoyu Ge
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Na Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yang Liu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Jing Huang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shixi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
13
|
Bárez‐López S, Bishop P, Searby D, Murphy D, Greenwood MP. Male rat hypothalamic extraretinal photoreceptor Opsin3 is sensitive to osmotic stimuli and light. J Neuroendocrinol 2024; 36:e13363. [PMID: 38192267 PMCID: PMC11475585 DOI: 10.1111/jne.13363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/13/2023] [Accepted: 12/16/2023] [Indexed: 01/10/2024]
Abstract
The light-sensitive protein Opsin 3 (Opn3) is present throughout the mammalian brain; however, the role of Opn3 in this organ remains unknown. Since Opn3 encoded mRNA is modulated in the supraoptic and paraventricular nucleus of the hypothalamus in response to osmotic stimuli, we have explored by in situ hybridization the expression of Opn3 in these nuclei. We have demonstrated that Opn3 is present in the male rat magnocellular neurones expressing either the arginine vasopressin or oxytocin neuropeptides and that Opn3 increases in both neuronal types in response to osmotic stimuli, suggesting that Opn3 functions in both cell types and that it might be involved in regulating water balance. Using rat hypothalamic organotypic cultures, we have demonstrated that the hypothalamus is sensitive to light and that the observed light sensitivity is mediated, at least in part, by Opn3. The data suggests that hypothalamic Opn3 can mediate a light-sensitive role to regulate circadian homeostatic processes.
Collapse
Affiliation(s)
- Soledad Bárez‐López
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
- Present address:
Instituto de Investigaciones BiomédicasConsejo Superior de Investigaciones Científicas (CSIC)—Universidad Autónoma de Madrid (UAM)MadridSpain
| | - Paul Bishop
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| | - Daniel Searby
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| | - David Murphy
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| | - Michael P. Greenwood
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| |
Collapse
|
14
|
Cero C, Shu W, Reese AL, Douglas D, Maddox M, Singh AP, Ali SL, Zhu AR, Katz JM, Pierce AE, Long KT, Nilubol N, Cypess RH, Jacobs JL, Tian F, Cypess AM. Standardized In Vitro Models of Human Adipose Tissue Reveal Metabolic Flexibility in Brown Adipocyte Thermogenesis. Endocrinology 2023; 164:bqad161. [PMID: 37944134 PMCID: PMC11032247 DOI: 10.1210/endocr/bqad161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/10/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Functional human brown and white adipose tissue (BAT and WAT) are vital for thermoregulation and nutritional homeostasis, while obesity and other stressors lead, respectively, to cold intolerance and metabolic disease. Understanding BAT and WAT physiology and dysfunction necessitates clinical trials complemented by mechanistic experiments at the cellular level. These require standardized in vitro models, currently lacking, that establish references for gene expression and function. We generated and characterized a pair of immortalized, clonal human brown (hBA) and white (hWA) preadipocytes derived from the perirenal and subcutaneous depots, respectively, of a 40-year-old male individual. Cells were immortalized with hTERT and confirmed to be of a mesenchymal, nonhematopoietic lineage based on fluorescence-activated cell sorting and DNA barcoding. Functional assessments showed that the hWA and hBA phenocopied primary adipocytes in terms of adrenergic signaling, lipolysis, and thermogenesis. Compared to hWA, hBA were metabolically distinct, with higher rates of glucose uptake and lactate metabolism, and greater basal, maximal, and nonmitochondrial respiration, providing a mechanistic explanation for the association between obesity and BAT dysfunction. The hBA also responded to the stress of maximal respiration by using both endogenous and exogenous fatty acids. In contrast to certain mouse models, hBA adrenergic thermogenesis was mediated by several mechanisms, not principally via uncoupling protein 1 (UCP1). Transcriptomics via RNA-seq were consistent with the functional studies and established a molecular signature for each cell type before and after differentiation. These standardized cells are anticipated to become a common resource for future physiological, pharmacological, and genetic studies of human adipocytes.
Collapse
Affiliation(s)
- Cheryl Cero
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiguo Shu
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Amy L Reese
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Diana Douglas
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Michael Maddox
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
- Current Affiliation: Vita Therapeutics, 801 W. Baltimore Street, Suite 301, Baltimore, MD 21201, USA
| | - Ajeet P Singh
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Sahara L Ali
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander R Zhu
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jacqueline M Katz
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne E Pierce
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kelly T Long
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naris Nilubol
- Surgical Oncology Program, Center for Cancer Research, NCI, NIH, 10 Center Drive, Room 4-5952, Bethesda, MD 20892, USA
| | - Raymond H Cypess
- American Type Culture Collection, 10801 University Blvd, Manassas, VA 20110, USA
| | - Jonathan L Jacobs
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Fang Tian
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
15
|
Andrabi M, Upton BA, Lang RA, Vemaraju S. An Expanding Role for Nonvisual Opsins in Extraocular Light Sensing Physiology. Annu Rev Vis Sci 2023; 9:245-267. [PMID: 37196422 DOI: 10.1146/annurev-vision-100820-094018] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
We live on a planet that is bathed in daily and seasonal sunlight cycles. In this context, terrestrial life forms have evolved mechanisms that directly harness light energy (plants) or decode light information for adaptive advantage. In animals, the main light sensors are a family of G protein-coupled receptors called opsins. Opsin function is best described for the visual sense. However, most animals also use opsins for extraocular light sensing for seasonal behavior and camouflage. While it has long been believed that mammals do not have an extraocular light sensing capacity, recent evidence suggests otherwise. Notably, encephalopsin (OPN3) and neuropsin (OPN5) are both known to mediate extraocular light sensing in mice. Examples of this mediation include photoentrainment of circadian clocks in skin (by OPN5) and acute light-dependent regulation of metabolic pathways (by OPN3 and OPN5). This review summarizes current findings in the expanding field of extraocular photoreception and their relevance for human physiology.
Collapse
Affiliation(s)
- Mutahar Andrabi
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; ,
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Brian A Upton
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; ,
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Molecular and Developmental Biology Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Medical Scientist Training Program, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Richard A Lang
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; ,
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Shruti Vemaraju
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; ,
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
16
|
Wang Y, Ye L. Somatosensory innervation of adipose tissues. Physiol Behav 2023; 265:114174. [PMID: 36965573 PMCID: PMC11537203 DOI: 10.1016/j.physbeh.2023.114174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
The increasing prevalence of obesity and type 2 diabetes has led to a greater interest in adipose tissue physiology. Adipose tissue is now understood as an organ with endocrine and thermogenic capacities in addition to its role in fat storage. It plays a critical role in systemic metabolism and energy regulation, and its activity is tightly regulated by the nervous system. Fat is now recognized to receive sympathetic innervation, which transmits information from the brain, as well as sensory innervation, which sends information into the brain. The role of sympathetic innervation in adipose tissue has been extensively studied. However, the extent and the functional significance of sensory innervation have long been unclear. Recent studies have started to reveal that sensory neurons robustly innervate adipose tissue and play an important role in regulating fat activity. This brief review will discuss both historical evidence and recent advances, as well as important remaining questions about the sensory innervation of adipose tissue.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
Linne C, Mon KY, D’Souza S, Jeong H, Jiang X, Brown DM, Zhang K, Vemaraju S, Tsubota K, Kurihara T, Pardue MT, Lang RA. Encephalopsin (OPN3) is required for normal refractive development and the GO/GROW response to induced myopia. Mol Vis 2023; 29:39-57. [PMID: 37287644 PMCID: PMC10243678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/05/2023] [Indexed: 06/09/2023] Open
Abstract
Purpose Myopia, or nearsightedness, is the most common form of refractive error and is increasing in prevalence. While significant efforts have been made to identify genetic variants that predispose individuals to myopia, these variants are believed to account for only a small portion of the myopia prevalence, leading to a feedback theory of emmetropization, which depends on the active perception of environmental visual cues. Consequently, there has been renewed interest in studying myopia in the context of light perception, beginning with the opsin family of G-protein coupled receptors (GPCRs). Refractive phenotypes have been characterized in every opsin signaling pathway studied, leaving only Opsin 3 (OPN3), the most widely expressed and blue-light sensing noncanonical opsin, to be investigated for function in the eye and refraction. Methods Opn3 expression was assessed in various ocular tissues using an Opn3eGFP reporter. Weekly refractive development in Opn3 retinal and germline mutants from 3 to 9 weeks of age was measured using an infrared photorefractor and spectral domain optical coherence tomography (SD-OCT). Susceptibility to lens-induced myopia was then assessed using skull-mounted goggles with a -30 diopter experimental and a 0 diopter control lens. Mouse eye biometry was similarly tracked from 3 to 6 weeks. A myopia gene expression signature was assessed 24 h after lens induction for germline mutants to further assess myopia-induced changes. Results Opn3 was found to be expressed in a subset of retinal ganglion cells and a limited number of choroidal cells. Based on an assessment of Opn3 mutants, the OPN3 germline, but not retina conditional Opn3 knockout, exhibits a refractive myopia phenotype, which manifests in decreased lens thickness, shallower aqueous compartment depth, and shorter axial length, atypical of traditional axial myopias. Despite the short axial length, Opn3 null eyes demonstrate normal axial elongation in response to myopia induction and mild changes in choroidal thinning and myopic shift, suggesting that susceptibility to lens-induced myopia is largely unchanged. Additionally, the Opn3 null retinal gene expression signature in response to induced myopia after 24 h is distinct, with opposing Ctgf, Cx43, and Egr1 polarity compared to controls. Conclusions The data suggest that an OPN3 expression domain outside the retina can control lens shape and thus the refractive performance of the eye. Prior to this study, the role of Opn3 in the eye had not been investigated. This work adds OPN3 to the list of opsin family GPCRs that are implicated in emmetropization and myopia. Further, the work to exclude retinal OPN3 as the contributing domain in this refractive phenotype is unique and suggests a distinct mechanism when compared to other opsins.
Collapse
Affiliation(s)
- Courtney Linne
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Khine Yin Mon
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Shane D’Souza
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Heonuk Jeong
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Xiaoyan Jiang
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Dillon M. Brown
- Department of Ophthalmology and Neuroscience Program, Emory University School of Medicine, Atlanta, GA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA
| | - Kevin Zhang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Shruti Vemaraju
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Tsubota Laboratory, Inc., Tokyo, Japan
| | - Toshihide Kurihara
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Machelle T. Pardue
- Department of Ophthalmology and Neuroscience Program, Emory University School of Medicine, Atlanta, GA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA
| | - Richard A. Lang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
18
|
Doucette CC, Nguyen DC, Barteselli D, Blanchard S, Pelletier M, Kesharwani D, Jachimowicz E, Su S, Karolak M, Brown AC. Optogenetic activation of UCP1-dependent thermogenesis in brown adipocytes. iScience 2023; 26:106560. [PMID: 37123235 PMCID: PMC10139976 DOI: 10.1016/j.isci.2023.106560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/01/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Brown adipocytes are unique in that they expend energy and produce heat to maintain euthermia through expression of uncoupling protein-1 (UCP1). Given their propensity to stimulate weight loss and promote resistance to obesity, they are a compelling interventional target for obesity-related disorders. Here, we tested whether an optogenetic approach could be used to activate UCP1-dependent thermogenesis in brown adipocytes. We generated brown adipocytes expressing a bacterial-derived photoactivatable adenylyl cyclase (bPAC) that, upon blue light stimulation, increases UCP1 expression, fuel uptake and thermogenesis. This unique system allows for precise, chemical free, temporal control of UCP1-dependent thermogenesis, which can aid in our understanding of brown adipocyte biology and development of therapies that target obesity-related disorders.
Collapse
Affiliation(s)
- Chad C. Doucette
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Daniel C. Nguyen
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Davide Barteselli
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Sophia Blanchard
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Masen Pelletier
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Devesh Kesharwani
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Ed Jachimowicz
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Su Su
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Michele Karolak
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Aaron C. Brown
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
- School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
- Tufts University School of Medicine, 145 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
19
|
Karthikeyan R, Davies WI, Gunhaga L. Non-image-forming functional roles of OPN3, OPN4 and OPN5 photopigments. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2023. [DOI: 10.1016/j.jpap.2023.100177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
|
20
|
GPCR in Adipose Tissue Function-Focus on Lipolysis. Biomedicines 2023; 11:biomedicines11020588. [PMID: 36831123 PMCID: PMC9953751 DOI: 10.3390/biomedicines11020588] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Adipose tissue can be divided anatomically, histologically, and functionally into two major entities white and brown adipose tissues (WAT and BAT, respectively). WAT is the primary energy depot, storing most of the bioavailable triacylglycerol molecules of the body, whereas BAT is designed for dissipating energy in the form of heat, a process also known as non-shivering thermogenesis as a defense against a cold environment. Importantly, BAT-dependent energy dissipation directly correlates with cardiometabolic health and has been postulated as an intriguing target for anti-obesity therapies. In general, adipose tissue (AT) lipid content is defined by lipid uptake and lipogenesis on one side, and, on the other side, it is defined by the breakdown of lipids and the release of fatty acids by lipolysis. The equilibrium between lipogenesis and lipolysis is important for adipocyte and general metabolic homeostasis. Overloading adipocytes with lipids causes cell stress, leading to the recruitment of immune cells and adipose tissue inflammation, which can affect the whole organism (metaflammation). The most important consequence of energy and lipid overload is obesity and associated pathophysiologies, including insulin resistance, type 2 diabetes, and cardiovascular disease. The fate of lipolysis products (fatty acids and glycerol) largely differs between AT: WAT releases fatty acids into the blood to deliver energy to other tissues (e.g., muscle). Activation of BAT, instead, liberates fatty acids that are used within brown adipocyte mitochondria for thermogenesis. The enzymes involved in lipolysis are tightly regulated by the second messenger cyclic adenosine monophosphate (cAMP), which is activated or inhibited by G protein-coupled receptors (GPCRs) that interact with heterotrimeric G proteins (G proteins). Thus, GPCRs are the upstream regulators of the equilibrium between lipogenesis and lipolysis. Moreover, GPCRs are of special pharmacological interest because about one third of the approved drugs target GPCRs. Here, we will discuss the effects of some of most studied as well as "novel" GPCRs and their ligands. We will review different facets of in vitro, ex vivo, and in vivo studies, obtained with both pharmacological and genetic approaches. Finally, we will report some possible therapeutic strategies to treat obesity employing GPCRs as primary target.
Collapse
|
21
|
Marchese NA, Ríos MN, Guido ME. Müller glial cell photosensitivity: a novel function bringing higher complexity to vertebrate retinal physiology. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2023. [DOI: 10.1016/j.jpap.2023.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
22
|
Koyanagi M, Honda H, Yokono H, Sato R, Nagata T, Terakita A. Expression of a homologue of a vertebrate non-visual opsin Opn3 in the insect photoreceptors. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210274. [PMID: 36058246 PMCID: PMC9441228 DOI: 10.1098/rstb.2021.0274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Insect vision starts with light absorption by visual pigments based on opsins that drive Gq-type G protein-mediated phototransduction. Since Drosophila, the most studied insect in vision research, has only Gq-coupled opsins, the Gq-mediated phototransduction has been solely focused on insect vision for decades. However, genome projects on mosquitos uncovered non-canonical insect opsin genes, members of the Opn3 or c-opsin group composed of vertebrate and invertebrate non-visual opsins. Here, we report that a homologue of Opn3, MosOpn3 (Asop12) is expressed in eyes of a mosquito Anopheles stephensi. In situ hybridization analysis revealed that MosOpn3 is expressed in dorsal and ventral ommatidia, in which only R7 photoreceptor cells express MosOpn3. We also found that Asop9, a Gq-coupled visual opsin, exhibited co-localization with MosOpn3. Spectroscopic analysis revealed that Asop9 forms a blue-sensitive opsin-based pigment. Thus, the Gi/Go-coupled opsin MosOpn3, which forms a green-sensitive pigment, is co-localized with Asop9, a Gq-coupled opsin that forms a blue-sensitive visual pigment. Since these two opsin-based pigments trigger different phototransduction cascades, the R7 photoreceptors could generate complex photoresponses to blue to green light. This article is part of the theme issue ‘Understanding colour vision: molecular, physiological, neuronal and behavioural studies in arthropods’.
Collapse
Affiliation(s)
- Mitsumasa Koyanagi
- Department of Biology, Graduate School of Science, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan.,Department of Biology and Geosciences, Graduate School of Science, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan.,The OCU Advanced Research Institute for Natural Science and Technology, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| | - Hayato Honda
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| | - Hirohisa Yokono
- Department of Earth and Space Science, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan
| | - Ryu Sato
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| | - Takashi Nagata
- Department of Biology and Geosciences, Graduate School of Science, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| | - Akihisa Terakita
- Department of Biology, Graduate School of Science, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan.,Department of Biology and Geosciences, Graduate School of Science, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan.,The OCU Advanced Research Institute for Natural Science and Technology, Osaka City University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka 558-8585, Japan
| |
Collapse
|
23
|
Comprehensive Behavioral Analysis of Opsin 3 (Encephalopsin)-Deficient Mice Identifies Role in Modulation of Acoustic Startle Reflex. eNeuro 2022; 9:ENEURO.0202-22.2022. [PMID: 36041828 PMCID: PMC9532019 DOI: 10.1523/eneuro.0202-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/13/2022] [Accepted: 06/23/2022] [Indexed: 12/15/2022] Open
Abstract
Opsin-3 (Opn3, encephalopsin) was the first nonvisual opsin gene discovered in mammals. Since then, several Opn3 functions have been described, and in two cases (adipose tissue, smooth muscle) light sensing activity is implicated. In addition to peripheral tissues, Opn3 is robustly expressed within the central nervous system, for which it derives its name. Despite this expression, no studies have investigated developmental or adult CNS consequences of Opn3 loss-of-function. Here, the behavioral consequences of mice deficient in Opn3 were investigated. Opn3-deficient mice perform comparably to wild-type mice in measures of motor coordination, socialization, anxiety-like behavior, and various aspects of learning and memory. However, Opn3-deficient mice have an attenuated acoustic startle reflex (ASR) relative to littermates. This deficit is not because of changes in hearing sensitivity, although Opn3 was shown to be expressed in auditory and vestibular structures, including cochlear outer hair cells. Interestingly, the ASR was not acutely light-dependent and did not vary between daytime and nighttime trials, despite known functions of Opn3 in photoreception and circadian gene amplitude. Together, these results demonstrate the first role of Opn3 on behavior, although the role of this opsin in the CNS remains largely elusive.
Collapse
|
24
|
Sugiyama Y, Shimokawa F, Sugiyama K, Kobayashi T, Yamashita Y, Kazama K, Onda K, Funaba M, Murakami M. Relationships between the expression of adipose genes and profiles of hospitalized dogs. Vet Res Commun 2022; 46:1239-1244. [PMID: 36048336 DOI: 10.1007/s11259-022-09989-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022]
Abstract
Obesity is one of the risk factors for the onset of various metabolic diseases in dogs. Energy expenditure in brown/beige adipocytes, which is partially regulated by the bone morphogenetic protein (BMP) pathway, is a key factor determining systemic energy balance. Here, we examined gene expression in the fat depots of 129 hospitalized dogs, and the relationship between the relative levels of gene expression and profiles of dogs. We evaluated the expression levels of 23 genes such as regulatory genes of adipocyte differentiation and function, adipokines, genes related to brown adipogenesis and uncoupling protein (Ucp), and genes involved in BMP signaling. A reliable equation of multiple regression was not obtained to explain the body condition score (BCS), which is an index of adiposity. Positive relationships were detected between the expression levels of many genes, except for Ucp1 or Ucp3. BCS was found to increase with age. BCS was negatively correlated to the expression levels of Pparγ and Fasn, and positively correlated to Leptin and Opn3 expression. Aging decreased the expression levels of genes related to adipocyte differentiation and function (Pparγ, Fabp4, Fasn, Hsl, and Insr) and Adipoq. In addition, age was negatively correlated with the expression of genes involved in brown adipogenesis and BMP signaling components (Prdm16, Bmp4, Alk3, Actr2a, and Actr2b). In contrast, the expression levels of Leptin and Ucp2 were found to increase with age. The present study clarifies BCS- and age-related gene expressions in the adipose tissue, which potentially contribute to elucidating the etiology of canine obesity.
Collapse
Affiliation(s)
- Yukina Sugiyama
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, 252-5201, Japan.,Sugiyama Animal Hospital, Shizuoka, 424-0068, Japan
| | - Fumie Shimokawa
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, 252-5201, Japan
| | - Kazutoshi Sugiyama
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, 252-5201, Japan.,Sugiyama Animal Hospital, Shizuoka, 424-0068, Japan
| | | | | | - Kei Kazama
- Laboratory of Farm Animal Internal Medicine, Azabu University School of Veterinary Medicine, Sagamihara, 252-5201, Japan
| | - Ken Onda
- Laboratory of Farm Animal Internal Medicine, Azabu University School of Veterinary Medicine, Sagamihara, 252-5201, Japan
| | - Masayuki Funaba
- Division of Applied Biosciences, Kyoto University Graduate School of Agriculture, Kyoto, 606-8502, Japan.
| | - Masaru Murakami
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, 252-5201, Japan.
| |
Collapse
|
25
|
Johansen OS, Ma T, Gerhart-Hines Z. Leveraging GPCR signaling in thermogenic fat to counteract metabolic diseases. Mol Metab 2022; 60:101474. [PMID: 35339729 PMCID: PMC9046952 DOI: 10.1016/j.molmet.2022.101474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/24/2022] [Accepted: 03/11/2022] [Indexed: 11/05/2022] Open
Abstract
Background Thermogenic brown and beige adipocytes are recognized for their unique capacity to consume extraordinary levels of metabolites and lipids from the blood to fuel heat-producing catabolic processes [[1], [2], [3], [4], [5], [6], [7]]. In humans, the functions of thermogenic adipocytes are associated with cardiometabolic protection and improved glycemic control [[8], [9], [10], [11], [12], [13]]. Consequently, engaging these macronutrient-consuming and energy-dissipating activities has gained attention as a promising therapeutic strategy for counteracting metabolic diseases, such as obesity and diabetes. Scope of review In this review, we highlight new advances in our understanding of the physiological role of G protein-coupled receptors (GPCRs) in controlling thermogenic adipocyte biology. We further extend our discussion to the opportunities and challenges posed by pharmacologically targeting different elements of GPCR signaling in these highly specialized fat cells. Major conclusions GPCRs represent appealing candidates through which to harness adipose thermogenesis. Yet safely and effectively targeting these druggable receptors on brown and beige adipocytes has thus far proven challenging. Therefore, continued interrogation across the GPCR landscape is necessary for future leaps within the field of thermogenic fat biology to unlock the therapeutic potential of adipocyte catabolism. Brown and beige thermogenic adipocytes robustly consume and catabolize macronutrients. The catabolic activity of thermogenic adipocytes promotes organismal energy balance. Thermogenic adipocyte functions are tightly controlled by G protein-coupled receptors (GPCRs). GPCRs can be potentially targeted at multiple levels to therapeutically harness thermogenic activity.
Collapse
Affiliation(s)
- Olivia Sveidahl Johansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK
| | - Tao Ma
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK; Embark Biotech ApS, Copenhagen, DK
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, DK; Embark Biotech ApS, Copenhagen, DK; Center for Adipocyte Signaling, Odense, DK.
| |
Collapse
|
26
|
Calligaro H, Dkhissi-Benyahya O, Panda S. Ocular and extraocular roles of neuropsin in vertebrates. Trends Neurosci 2022; 45:200-211. [PMID: 34952723 PMCID: PMC8854378 DOI: 10.1016/j.tins.2021.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/04/2021] [Accepted: 11/23/2021] [Indexed: 01/13/2023]
Abstract
The ability to detect and adapt to different levels of ambient light is critical for animal survival. Light detection is the basis of vision, but light also regulates eye development and drives several non-image-forming functions, including synchronizing circadian rhythms to the daily light/dark cycle, restricting pupils in response to changes in light intensity, and modulating mood in response to light. Until the early 2000s, these functions were thought to be solely mediated by ocular photoreceptors. However, neuropsin (OPN5), a UV-sensitive opsin, has been receiving growing attention, as new methods have revealed previously unappreciated functions of OPN5. In fact, OPN5-mediated extraocular and deep-brain photoreception have recently been described for the first time in mammals. This review aims to synthesize current knowledge of the properties and functions of OPN5 across vertebrates.
Collapse
Affiliation(s)
- Hugo Calligaro
- Regulatory Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ouria Dkhissi-Benyahya
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| | - Satchidananda Panda
- Regulatory Biology, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
27
|
Liebert A, Pang V, Bicknell B, McLachlan C, Mitrofanis J, Kiat H. A Perspective on the Potential of Opsins as an Integral Mechanism of Photobiomodulation: It's Not Just the Eyes. Photobiomodul Photomed Laser Surg 2022; 40:123-135. [PMID: 34935507 DOI: 10.1089/photob.2021.0106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective: To investigate the potential relationship between opsins and photobiomodulation. Background: Opsins and other photoreceptors occur in all phyla and are important in light-activated signaling and organism homeostasis. In addition to the visual opsin systems of the retina (OPN1 and OPN2), there are several non-visual opsins found throughout the body tissues, including encephalopsin/panopsin (OPN3), melanopsin (OPN4), and neuropsin (OPN5), as well as other structures that have light-sensitive properties, such as enzymes, ion channels, particularly those located in cell membranes, lysosomes, and neuronal structures such as the nodes of Ranvier. The influence of these structures on exposure to light, including self-generated light within the body (autofluorescence), on circadian oscillators, and circadian and ultradian rhythms have become increasingly reported. The visual and non-visual phototransduction cascade originating from opsins and other structures has potential significant mechanistic effects on tissues and health. Methods: A PubMed and Google Scholar search was made using the search terms "photobiomodulation", "light", "neuron", "opsins", "neuropsin", "melanopsin", "encephalopsin", "rhodopsin", and "chromophore". Results: This review was examined the influence of neuropsin (also known as kallikrein 8), encephalopsin, and melanopsin specifically on ion channel function, and more broadly on the central and peripheral nervous systems. The relationship between opsins 3, 4, and 5 and photobiomodulation mechanisms was evaluated, along with a proposed role of photobiomodulation through opsins and light-sensitive organelles as potential alleviators of symptoms and accelerators of beneficial regenerative processes. The potential clinical implications of this in musculoskeletal conditions, wounds, and in the symptomatic management of neurodegenerative disease was also examined. Conclusions: Systematic research into the pleotropic therapeutic role of photobiomodulation, mediated through its action on opsins and other light-sensitive organelles may assist in the future execution of safe, low-risk precision medicine for a variety of chronic and complex disease conditions, and for health maintenance in aging.
Collapse
Affiliation(s)
- Ann Liebert
- Faculty of Medicine and Health Sciences, University of Sydney, Sydney, Australia.,Office of Governance and Research, San Hospital, Sydney, Australia
| | | | - Brian Bicknell
- Faculty of Health Science, Australian Catholic University, North Sydney, Australia
| | | | - John Mitrofanis
- Clinatec, Fonds de Dotation-CEA, Universitè Grenoble Alpes, Grenoble, France
| | - Hosen Kiat
- Department of Clinical Medicine, Macquarie University, Sydney, Australia.,Cardiac Health Institute, Sydney, Australia
| |
Collapse
|
28
|
Ekechukwu ON, Christian M. Metabolic responses of light and taste receptors - unexpected actions of GPCRs in adipocytes. Rev Endocr Metab Disord 2022; 23:111-120. [PMID: 34195966 PMCID: PMC8873064 DOI: 10.1007/s11154-021-09667-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2021] [Indexed: 11/30/2022]
Abstract
The G-protein-coupled receptor (GPCR) superfamily includes sensory receptors that can detect and respond to taste and light. Recent investigations have identified key metabolic roles for such receptors in tissues considered 'non-sensory' such as adipose tissue. The major functions of white and brown adipose tissues include energy storage/release and thermogenesis, respectively. These processes are tightly controlled by GPCR pathways that serve to maintain energy homeostasis. Opsins 3 and 4 are GPCRs activated by blue light and in adipocytes control lipolysis as well as affect brown adipocyte activity. Furthermore, Opsin 3 signals to regulate the conversion of white to thermogenic beige/BRITE (Brown-in-white) adipocytes. Taste receptors that respond to fatty acids, sweet and bitter are expressed in adipocytes as well as in taste buds. Ffar2 and the long chain fatty acid receptor GPR120 are highly expressed in white adipocytes and the human tongue. In adipose tissue Ffar2 mediates the metabolic effects of butyrate and propionate produced by the gut microbiome. GPR120 is highly expressed in brown adipose tissue and regulates fatty acid oxidation and mitochondrial function. The type I taste receptor Tas1r3 senses sweet and umami, is expressed in adipocytes and on obesogenic diets Tas1r3 global gene knockout protects from metabolic dysfunction. Type II taste receptors that sense bitter are expressed by adipocytes and bitter agonists have been found to modulate adipocyte differentiation and lipid storage levels. This review explores recent unexpected findings of light and taste receptors in adipocytes and examines effects of their signaling in the control of adipose tissue biology.
Collapse
Affiliation(s)
- Onyinye Nuella Ekechukwu
- School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, UK
| | - Mark Christian
- School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, UK.
| |
Collapse
|
29
|
Marchese NA, Ríos MN, Guido ME. The Intrinsic Blue Light Responses of Avian Müller Glial Cells Imply Calcium Release from Internal Stores. ASN Neuro 2022; 14:17590914221076698. [PMID: 35103506 PMCID: PMC8814826 DOI: 10.1177/17590914221076698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The retina of vertebrates is responsible for capturing light through visual
(cones and rods) and non-visual photoreceptors (intrinsically photosensitive
retinal ganglion cells and horizontal cells) triggering a number of essential
activities associated to image- and non-image forming functions (photic
entrainment of daily rhythms, pupillary light reflexes, pineal melatonin
inhibition, among others). Although the retina contains diverse types of
neuronal based-photoreceptors cells, originally classified as ciliary- or
rhabdomeric-like types, in recent years, it has been shown that the major glial
cell type of the retina, the Müller glial cells (MC), express blue photopigments
as Opn3 (encephalopsin) and Opn5 (neuropsin) and display light responses
associated to intracellular Ca2 + mobilization. These findings strongly propose
MC as novel retinal photodetectors (Rios et al., 2019). Herein, we further
investigated the intrinsic light responses of primary cultures of MC from
embryonic chicken retinas specially focused on Ca2 + mobilization by
fluorescence imaging and the identity of the internal Ca2 + stores responsible
for blue light responses. Results clearly demonstrated that light responses were
specific to blue light of long time exposure, and that the main Ca2 + reservoir
to trigger downstream responses came from intracellular stores localized in the
endoplasmic reticulum These observations bring more complexity to the intrinsic
photosensitivity of retinal cells, particularly with regard to the detection of
light in the blue range of visible spectra, and add novel functions to glial
cells cooperating with other photoreceptors to detect and integrate ambient
light in the retinal circuit and participate in cell to cell communication.
Summary statement:
Non-neuronal cells in the vertebrate retina, Muller glial cells, express
non-canonical photopigments and sense blue light causing calcium release from
intracellular stores strongly suggesting a novel intrinsic photosensitivity and
new regulatory events mediating light-driven processes with yet unknown
physiological implications.
Collapse
Affiliation(s)
- Natalia A Marchese
- 373607CIQUIBIC-CONICET, Facultad de Ciencias Químicas, 28217Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maximiliano N Ríos
- 373607CIQUIBIC-CONICET, Facultad de Ciencias Químicas, 28217Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mario E Guido
- 373607CIQUIBIC-CONICET, Facultad de Ciencias Químicas, 28217Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
30
|
Abstract
Rhodopsins are photoreceptive membrane proteins consisting of a common heptahelical transmembrane architecture that contains a retinal chromophore. Rhodopsin was first discovered in the animal retina in 1876, but a different type of rhodopsin, bacteriorhodopsin, was reported to be present in the cell membrane of an extreme halophilic archaeon, Halobacterium salinarum, 95 years later. Although these findings were made by physiological observation of pigmented tissue and cell bodies, recent progress in genomic and metagenomic analyses has revealed that there are more than 10,000 microbial rhodopsins and 9000 animal rhodopsins with large diversity and tremendous new functionality. In this Cell Science at a Glance article and accompanying poster, we provide an overview of the diversity of functions, structures, color discrimination mechanisms and optogenetic applications of these two rhodopsin families, and will also highlight the third distinctive rhodopsin family, heliorhodopsin.
Collapse
Affiliation(s)
- Takashi Nagata
- The Institute for Solid State Physics, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8581, Japan.,PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Keiichi Inoue
- The Institute for Solid State Physics, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8581, Japan
| |
Collapse
|
31
|
Nakano T, Chiang KC, Chen CC, Chen PJ, Lai CY, Hsu LW, Ohmori N, Goto T, Chen CL, Goto S. Sunlight Exposure and Phototherapy: Perspectives for Healthy Aging in an Era of COVID-19. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182010950. [PMID: 34682694 PMCID: PMC8535353 DOI: 10.3390/ijerph182010950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 01/10/2023]
Abstract
Most humans depend on sunlight exposure to satisfy their requirements for vitamin D3. However, the destruction of the ozone layer in the past few decades has increased the risk of skin aging and wrinkling caused by excessive exposure to ultraviolet (UV) radiation, which may also promote the risk of skin cancer development. The promotion of public health recommendations to avoid sunlight exposure would reduce the risk of skin cancer, but it would also enhance the risk of vitamin D3 insufficiency/deficiency, which may cause disease development and progression. In addition, the ongoing global COVID-19 pandemic may further reduce sunlight exposure due to stay-at-home policies, resulting in difficulty in active and healthy aging. In this review article, we performed a literature search in PubMed and provided an overview of basic and clinical data regarding the impact of sunlight exposure and vitamin D3 on public health. We also discuss the potential mechanisms and clinical value of phototherapy with a full-spectrum light (notably blue, red, and near-infrared light) as an alternative to sunlight exposure, which may contribute to combating COVID-19 and promoting active and healthy aging in current aged/superaged societies.
Collapse
Affiliation(s)
- Toshiaki Nakano
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (K.-C.C.); (P.-J.C.); (L.-W.H.)
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-C.C.); (C.-Y.L.); (C.-L.C.)
- Correspondence: (T.N.); (S.G.); Tel.: +886-7-731-7123 (T.N.); +81-975-53-2165 (S.G.)
| | - Kuei-Chen Chiang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (K.-C.C.); (P.-J.C.); (L.-W.H.)
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-C.C.); (C.-Y.L.); (C.-L.C.)
| | - Chien-Chih Chen
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-C.C.); (C.-Y.L.); (C.-L.C.)
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Po-Jung Chen
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (K.-C.C.); (P.-J.C.); (L.-W.H.)
| | - Chia-Yun Lai
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-C.C.); (C.-Y.L.); (C.-L.C.)
| | - Li-Wen Hsu
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (K.-C.C.); (P.-J.C.); (L.-W.H.)
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-C.C.); (C.-Y.L.); (C.-L.C.)
| | - Naoya Ohmori
- Faculty of Nursing, Josai International University, Togane 283-8555, Japan; (N.O.); (T.G.)
- Kazusa Institute for Drug Discovery, Josai International University, Togane 283-8555, Japan
| | - Takeshi Goto
- Faculty of Nursing, Josai International University, Togane 283-8555, Japan; (N.O.); (T.G.)
- Kazusa Institute for Drug Discovery, Josai International University, Togane 283-8555, Japan
| | - Chao-Long Chen
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-C.C.); (C.-Y.L.); (C.-L.C.)
| | - Shigeru Goto
- Liver Transplantation Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan; (C.-C.C.); (C.-Y.L.); (C.-L.C.)
- Faculty of Nursing, Josai International University, Togane 283-8555, Japan; (N.O.); (T.G.)
- Nobeoka Medical Check Center, Fukuoka Institution of Occupational Health, Nobeoka 882-0872, Japan
- Correspondence: (T.N.); (S.G.); Tel.: +886-7-731-7123 (T.N.); +81-975-53-2165 (S.G.)
| |
Collapse
|
32
|
Kojima K. [Biophysical and Biochemical Research of Animal Rhodopsins]. YAKUGAKU ZASSHI 2021; 141:1155-1160. [PMID: 34602512 DOI: 10.1248/yakushi.21-00144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Opsins (also called animal rhodopsins) are universal photoreceptive proteins that provide the molecular basis of visual and nonvisual photoreception in animals, including humans. Opsins consist of seven helical α-transmembrane domains and use retinal, a derivative of vitamin A, as a chromophore. In many opsins, light absorption triggers photo-isomerization from 11-cis retinal to all-trans retinal, resulting in activation via dynamic structural changes in the protein moiety. Activated opsins stimulate cognate trimeric G proteins to induce signal transduction cascades in cells. Recently, molecular and physiological analyses of diverse opsins have progressively advanced. This review introduces the molecular basis and physiological functions of opsins. Based on the functions of opsins, I will discuss the potential of opsins as target molecules to treat and prevent visual and nonvisual diseases such as sleep disorder and depression.
Collapse
Affiliation(s)
- Keiichi Kojima
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| |
Collapse
|
33
|
Levine DC, Ramsey KM, Bass J. Circadian NAD(P)(H) cycles in cell metabolism. Semin Cell Dev Biol 2021; 126:15-26. [PMID: 34281771 DOI: 10.1016/j.semcdb.2021.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
Intrinsic circadian clocks are present in all forms of photosensitive life, enabling daily anticipation of the light/dark cycle and separation of energy storage and utilization cycles on a 24-h timescale. The core mechanism underlying circadian rhythmicity involves a cell-autonomous transcription/translation feedback loop that in turn drives rhythmic organismal physiology. In mammals, genetic studies have established that the core clock plays an essential role in maintaining metabolic health through actions within both brain pacemaker neurons and peripheral tissues and that disruption of the clock contributes to disease. Peripheral clocks, in turn, can be entrained by metabolic cues. In this review, we focus on the role of the nucleotide NAD(P)(H) and NAD+-dependent sirtuin deacetylases as integrators of circadian and metabolic cycles, as well as the implications for this interrelationship in healthful aging.
Collapse
Affiliation(s)
- Daniel C Levine
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kathryn M Ramsey
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
34
|
Opsins outside the eye and the skin: a more complex scenario than originally thought for a classical light sensor. Cell Tissue Res 2021; 385:519-538. [PMID: 34236517 DOI: 10.1007/s00441-021-03500-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/23/2021] [Indexed: 12/19/2022]
Abstract
Since the discovery of melanopsin as a retinal non-visual photopigment, opsins have been described in several organs and cells. This distribution is strikingly different from the classical localization of photopigments in light-exposed tissues such as the eyes and the skin. More than 10 years ago, a new paradigm in the field was created as opsins were shown, to detect not only light, but also thermal energy in Drosophila. In agreement with these findings, thermal detection by opsins was also reported in mammalian cells. Considering the presence of opsins in tissues not reached by light, an intriguing question has emerged: What is the role of a classical light-sensor, and more recently appreciated thermo-sensor, in these tissues? To tackle this question, we address in this review the most recent studies in the field, with emphasis in mammals. We provide the present view about the role of opsins in peripheral tissues, aiming to integrate the current knowledge of the presence and function of opsins in organs that are not directly affected by light.
Collapse
|
35
|
Poll BG, Chen L, Chou CL, Raghuram V, Knepper MA. Landscape of GPCR expression along the mouse nephron. Am J Physiol Renal Physiol 2021; 321:F50-F68. [PMID: 34029142 PMCID: PMC8321805 DOI: 10.1152/ajprenal.00077.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022] Open
Abstract
Kidney transport and other renal functions are regulated by multiple G protein-coupled receptors (GPCRs) expressed along the renal tubule. The rapid, recent appearance of comprehensive unbiased gene expression data in the various renal tubule segments, chiefly RNA sequencing and protein mass spectrometry data, has provided a means of identifying patterns of GPCR expression along the renal tubule. To allow for comprehensive mapping, we first curated a comprehensive list of GPCRs in the genomes of mice, rats, and humans (https://hpcwebapps.cit.nih.gov/ESBL/Database/GPCRs/) using multiple online data sources. We used this list to mine segment-specific and cell type-specific expression data from RNA-sequencing studies in microdissected mouse tubule segments to identify GPCRs that are selectively expressed in discrete tubule segments. Comparisons of these mapped mouse GPCRs with other omics datasets as well as functional data from isolated perfused tubule and micropuncture studies confirmed patterns of expression for well-known receptors and identified poorly studied GPCRs that are likely to play roles in the regulation of renal tubule function. Thus, we provide data resources for GPCR expression across the renal tubule, highlighting both well-known GPCRs and understudied receptors to provide guidance for future studies.
Collapse
Affiliation(s)
- Brian G Poll
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lihe Chen
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Chung-Lin Chou
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Viswanathan Raghuram
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
36
|
Chen Z, Huang S, Liu M. The review of the light parameters and mechanisms of Photobiomodulation on melanoma cells. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2021; 38:3-11. [PMID: 34181781 DOI: 10.1111/phpp.12715] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 05/27/2021] [Accepted: 06/25/2021] [Indexed: 11/30/2022]
Abstract
Photobiomodulation (PBM) uses low-intensity visible or near-infrared light to produce beneficial effects on cells or tissues, such as brain therapy, wound healing. Still there is no consistent recommendation on the parameters (dose, light mode, wavelength, irradiance) and protocols (repetition, treatment duration) for its clinical application. Herein, we summarize the current PBM parameters for the treatment of melanoma, and we also discuss the potential photoreceptors and downstream signaling mechanisms in the PBM treatment of melanoma cells. It is hypothesized that PBM may inhibit the melanoma cells by activating mitochondria, OPNs, and other receptors. Regardless of the underlying mechanisms, PBM has been shown to be beneficial in treating melanoma. Through further in-depth studies of the underlying potential mechanisms, it can strengthen the applications of PBM for the therapy of melanoma.
Collapse
Affiliation(s)
- Zeqing Chen
- Institute of Future Lighting, Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Shijie Huang
- Zhongshan Fudan Joint Innovation Center, Zhongshan City, China.,Institute for Electric Light Sources, Fudan University, Shanghai, China
| | - Muqing Liu
- Institute of Future Lighting, Academy for Engineering and Technology, Fudan University, Shanghai, China.,Zhongshan Fudan Joint Innovation Center, Zhongshan City, China.,Institute for Electric Light Sources, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Upton BA, Díaz NM, Gordon SA, Van Gelder RN, Buhr ED, Lang RA. Evolutionary Constraint on Visual and Nonvisual Mammalian Opsins. J Biol Rhythms 2021; 36:109-126. [PMID: 33765865 PMCID: PMC8058843 DOI: 10.1177/0748730421999870] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Animals have evolved light-sensitive G protein-coupled receptors, known as opsins, to detect coherent and ambient light for visual and nonvisual functions. These opsins have evolved to satisfy the particular lighting niches of the organisms that express them. While many unique patterns of evolution have been identified in mammals for rod and cone opsins, far less is known about the atypical mammalian opsins. Using genomic data from over 400 mammalian species from 22 orders, unique patterns of evolution for each mammalian opsins were identified, including photoisomerases, RGR-opsin (RGR) and peropsin (RRH), as well as atypical opsins, encephalopsin (OPN3), melanopsin (OPN4), and neuropsin (OPN5). The results demonstrate that OPN5 and rhodopsin show extreme conservation across all mammalian lineages. The cone opsins, SWS1 and LWS, and the nonvisual opsins, OPN3 and RRH, demonstrate a moderate degree of sequence conservation relative to other opsins, with some instances of lineage-specific gene loss. Finally, the photoisomerase, RGR, and the best-studied atypical opsin, OPN4, have high sequence diversity within mammals. These conservation patterns are maintained in human populations. Importantly, all mammalian opsins retain key amino acid residues important for conjugation to retinal-based chromophores, permitting light sensitivity. These patterns of evolution are discussed along with known functions of each atypical opsin, such as in circadian or metabolic physiology, to provide insight into the observed patterns of evolutionary constraint.
Collapse
Affiliation(s)
- Brian A. Upton
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Molecular & Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Nicolás M. Díaz
- Department of Ophthalmology, University of Washington School of Medicine, Seattle, Washington
| | - Shannon A. Gordon
- Department of Ophthalmology, University of Washington School of Medicine, Seattle, Washington
| | - Russell N. Van Gelder
- Department of Ophthalmology, University of Washington School of Medicine, Seattle, Washington
- Departments of Biological Structure and Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Ethan D. Buhr
- Department of Ophthalmology, University of Washington School of Medicine, Seattle, Washington
| | - Richard A. Lang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Ophthalmology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
38
|
Heyde I, Begemann K, Oster H. Contributions of white and brown adipose tissues to the circadian regulation of energy metabolism. Endocrinology 2021; 162:6102571. [PMID: 33453099 PMCID: PMC7864004 DOI: 10.1210/endocr/bqab009] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/17/2022]
Abstract
The term energy metabolism comprises the entirety of chemical processes associated with uptake, conversion, storage, and breakdown of nutrients. All these must be tightly regulated in time and space to ensure metabolic homeostasis in an environment characterized by cycles such as the succession of day and night. Most organisms evolved endogenous circadian clocks to achieve this goal. In mammals, a ubiquitous network of cellular clocks is coordinated by a pacemaker residing in the hypothalamic suprachiasmatic nucleus. Adipocytes harbor their own circadian clocks, and large aspects of adipose physiology are regulated in a circadian manner through transcriptional regulation of clock-controlled genes. White adipose tissue (WAT) stores energy in the form of triglycerides at times of high energy levels that then serve as fuel in times of need. It also functions as an endocrine organ, releasing factors in a circadian manner to regulate food intake and energy turnover in other tissues. Brown adipose tissue (BAT) produces heat through nonshivering thermogenesis, a process also controlled by the circadian clock. We here review how WAT and BAT contribute to the circadian regulation of energy metabolism. We describe how adipose rhythms are regulated by the interplay of systemic signals and local clocks and summarize how adipose-originating circadian factors feed-back on metabolic homeostasis. The role of adipose tissue in the circadian control of metabolism becomes increasingly clear as circadian disruption leads to alterations in adipose tissue regulation, promoting obesity and its sequelae. Stabilizing adipose tissue rhythms, in turn, may help to combat disrupted energy homeostasis and obesity.
Collapse
Affiliation(s)
- Isabel Heyde
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | | | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
- Correspondence: Henrik Oster, PhD, Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
39
|
Díaz NM, Lang RA, Van Gelder RN, Buhr ED. Wounding Induces Facultative Opn5-Dependent Circadian Photoreception in the Murine Cornea. Invest Ophthalmol Vis Sci 2021; 61:37. [PMID: 32543667 PMCID: PMC7415322 DOI: 10.1167/iovs.61.6.37] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Autonomous molecular circadian clocks are present in the majority of mammalian tissues. These clocks are synchronized to phases appropriate for their physiologic role by internal systemic cues, external environmental cues, or both. The circadian clocks of the in vivo mouse cornea synchronize to the phase of the brain's master clock primarily through systemic cues, but ex vivo corneal clocks entrain to environmental light cycles. We evaluated the underlying mechanisms of this difference. Methods Molecular circadian clocks of mouse corneas were evaluated in vivo and ex vivo for response to environmental light. The presence of opsins and effect of genetic deletion of opsins were evaluated for influence on circadian photoresponses. Opn5-expressing cells were identified using Opn5Cre;Ai14 mice and RT-PCR, and they were characterized using immunocytochemistry. Results Molecular circadian clocks of the cornea remain in phase with behavioral circadian locomotor rhythms in vivo but are photoentrainable in tissue culture. After full-thickness incision or epithelial debridement, expression of the opsin photopigment Opn5 is induced in the cornea in a subset of preexisting epithelial cells adjacent to the wound site. This induction coincides with conferral of direct, short-wavelength light sensitivity to the circadian clocks throughout the cornea. Conclusions Corneal circadian rhythms become photosensitive after wounding. Opn5 gene function (but not Opn3 or Opn4 function) is necessary for induced photosensitivity. These results demonstrate that opsin-dependent direct light sensitivity can be facultatively induced in the murine cornea.
Collapse
|
40
|
Kurtz R, Steinberg LG, Betcher M, Fowler D, Shepard BD. The Sensing Liver: Localization and Ligands for Hepatic Murine Olfactory and Taste Receptors. Front Physiol 2020; 11:574082. [PMID: 33123030 PMCID: PMC7573564 DOI: 10.3389/fphys.2020.574082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/15/2020] [Indexed: 11/13/2022] Open
Abstract
Sensory receptors, including olfactory receptors (ORs), taste receptors (TRs), and opsins (Opns) have recently been found in a variety of non-sensory tissues where they have distinct physiological functions. As G protein-coupled receptors (GPCRs), these proteins can serve as important chemosensors by sensing and interpreting chemical cues in the environment. We reasoned that the liver, the largest metabolic organ in the body, is primed to take advantage of some of these sensory receptors in order to sense and regulate blood content and metabolism. In this study, we report the expression of novel hepatic sensory receptors - including 7 ORs, 6 bitter TRs, and 1 Opn - identified through a systematic molecular biology screening approach. We further determined that several of these receptors are expressed within hepatocytes, the parenchymal cells of the liver. Finally, we uncovered several agonists of the previously orphaned hepatic ORs. These compounds fall under two classes: methylpyrazines and monoterpenes. In particular, the latter chemicals are plant and fungal-derived compounds with known hepatic protective effects. Collectively, this study sheds light on the chemosensory functions of the liver and unveils potentially important regulators of hepatic homeostasis.
Collapse
Affiliation(s)
- Ryan Kurtz
- Department of Human Science, Georgetown University, Washington, DC, United States
| | - Lily G Steinberg
- Department of Human Science, Georgetown University, Washington, DC, United States
| | - Madison Betcher
- Department of Human Science, Georgetown University, Washington, DC, United States
| | - Dalton Fowler
- Department of Human Science, Georgetown University, Washington, DC, United States
| | - Blythe D Shepard
- Department of Human Science, Georgetown University, Washington, DC, United States
| |
Collapse
|
41
|
|
42
|
Olinski LE, Tsuda AC, Kauer JA, Oancea E. Endogenous Opsin 3 (OPN3) Protein Expression in the Adult Brain Using a Novel OPN3-mCherry Knock-In Mouse Model. eNeuro 2020; 7:ENEURO.0107-20.2020. [PMID: 32737180 PMCID: PMC7477952 DOI: 10.1523/eneuro.0107-20.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 01/07/2023] Open
Abstract
The opsins have been studied extensively for their functions in visual phototransduction; however, the mechanisms underlying extraocular opsin signaling remain poorly understood. The first mammalian extraocular opsin to be discovered, opsin 3 (OPN3), was found in the brain more than two decades ago, yet its function remains unknown. A significant hindrance to studying OPN3 has been a lack of specific antibodies against mammalian OPN3, resulting in an incomplete understanding of its expression in the brain. Although Opn3 promoter-driven reporter mice have been generated to examine general OPN3 localization, they lack the regulated expression of the endogenous protein and the ability to study its subcellular localization. To circumvent these issues, we have created a knock-in OPN3-mCherry mouse model in which the fusion protein OPN3-mCherry is expressed under the endogenous Opn3 promoter. Viable and fertile homozygotes for the OPN3-mCherry allele were used to create an extensive map of OPN3-mCherry expression across the adult mouse brain. OPN3-mCherry was readily visualized in distinct layers of the cerebral cortex (CTX), the hippocampal formation (HCF), distinct nuclei of the thalamus, as well as many other regions in both neuronal and non-neuronal cells. Our mouse model offers a new platform to investigate the function of OPN3 in the brain.
Collapse
Affiliation(s)
- Lauren E Olinski
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912
| | - Ayumi C Tsuda
- Department of Molecular Pharmacology and Physiology, Brown University, Providence, RI 02912
| | - Julie A Kauer
- Department of Psychiatry, Stanford University, Stanford, CA 94305
| | - Elena Oancea
- Department of Molecular Pharmacology and Physiology, Brown University, Providence, RI 02912
| |
Collapse
|
43
|
Zhang KX, D'Souza S, Upton BA, Kernodle S, Vemaraju S, Nayak G, Gaitonde KD, Holt AL, Linne CD, Smith AN, Petts NT, Batie M, Mukherjee R, Tiwari D, Buhr ED, Van Gelder RN, Gross C, Sweeney A, Sanchez-Gurmaches J, Seeley RJ, Lang RA. Violet-light suppression of thermogenesis by opsin 5 hypothalamic neurons. Nature 2020; 585:420-425. [PMID: 32879486 PMCID: PMC8130195 DOI: 10.1038/s41586-020-2683-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022]
Abstract
The opsin family of G-protein-coupled receptors are used as light detectors in animals. Opsin 5 (also known as neuropsin or OPN5) is a highly conserved opsin that is sensitive to visible violet light1,2. In mice, OPN5 is a known photoreceptor in the retina3 and skin4 but is also expressed in the hypothalamic preoptic area (POA)5. Here we describe a light-sensing pathway in which POA neurons that express Opn5 regulate thermogenesis in brown adipose tissue (BAT). We show that Opn5 is expressed in glutamatergic warm-sensing POA neurons that receive synaptic input from several thermoregulatory nuclei. We further show that Opn5 POA neurons project to BAT and decrease its activity under chemogenetic stimulation. Opn5-null mice show overactive BAT, increased body temperature, and exaggerated thermogenesis when cold-challenged. Moreover, violet photostimulation during cold exposure acutely suppresses BAT temperature in wild-type mice but not in Opn5-null mice. Direct measurements of intracellular cAMP ex vivo show that Opn5 POA neurons increase cAMP when stimulated with violet light. This analysis thus identifies a violet light-sensitive deep brain photoreceptor that normally suppresses BAT thermogenesis.
Collapse
Affiliation(s)
- Kevin X Zhang
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Shane D'Souza
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Brian A Upton
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Stace Kernodle
- Department of Surgery, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Shruti Vemaraju
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gowri Nayak
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kevin D Gaitonde
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Amanda L Holt
- Department of Physics, Yale University, New Haven, CT, USA
| | - Courtney D Linne
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - April N Smith
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nathan T Petts
- Division of Clinical Engineering, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew Batie
- Division of Clinical Engineering, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rajib Mukherjee
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Durgesh Tiwari
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ethan D Buhr
- Department of Ophthalmology, University of Washington Medical School, Seattle, WA, USA
| | - Russell N Van Gelder
- Department of Ophthalmology, University of Washington Medical School, Seattle, WA, USA
- Department of Biological Structure, University of Washington Medical School, Seattle, WA, USA
- Department of Pathology, University of Washington Medical School, Seattle, WA, USA
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Alison Sweeney
- Department of Physics, Yale University, New Haven, CT, USA
| | - Joan Sanchez-Gurmaches
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, School of Public Health, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Richard A Lang
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Center for Chronobiology, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Ophthalmology, University of Cincinnati, College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
44
|
Cell-autonomous light sensitivity via Opsin3 regulates fuel utilization in brown adipocytes. PLoS Biol 2020; 18:e3000630. [PMID: 32040503 PMCID: PMC7034924 DOI: 10.1371/journal.pbio.3000630] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/21/2020] [Accepted: 01/27/2020] [Indexed: 01/15/2023] Open
Abstract
Opsin3 (Opn3) is a transmembrane heptahelical G protein–coupled receptor (GPCR) with the potential to produce a nonvisual photoreceptive effect. Interestingly, anatomical profiling of GPCRs reveals that Opn3 mRNA is highly expressed in adipose tissue. The photosensitive functions of Opn3 in mammals are poorly understood, and whether Opn3 has a role in fat is entirely unknown. In this study, we found that Opn3-knockout (Opn3-KO) mice were prone to diet-induced obesity and insulin resistance. At the cellular level, Opn3-KO brown adipocytes cultured in darkness had decreased glucose uptake and lower nutrient-induced mitochondrial respiration than wild-type (WT) cells. Light exposure promoted mitochondrial activity and glucose uptake in WT adipocytes but not in Opn3-KO cells. Brown adipocytes carrying a defective mutation in Opn3’s putative G protein–binding domain also exhibited a reduction in glucose uptake and mitochondrial respiration in darkness. Using RNA-sequencing, we identified several novel light-sensitive and Opn3-dependent molecular signatures in brown adipocytes. Importantly, direct exposure of brown adipose tissue (BAT) to light in living mice significantly enhanced thermogenic capacity of BAT, and this effect was diminished in Opn3-KO animals. These results uncover a previously unrecognized cell-autonomous, light-sensing mechanism in brown adipocytes via Opn3-GPCR signaling that can regulate fuel metabolism and mitochondrial respiration. Our work also provides a molecular basis for developing light-based treatments for obesity and its related metabolic disorders. Brown adipose tissue plays a pivotal role in energy homeostasis and serves as a metabolic sink for glucose and fatty acid. This study demonstrates a novel light-sensing mechanism in mice via the photoreceptor Opsin3 that regulates fuel utilization and mitochondrial activity of brown adipocytes.
Collapse
|